1
|
Sohrabi S, Alipour S, Ghahramanipour Z, Masoumi J, Baradaran B. STAT signaling pathways in immune cells and their associated mechanisms in cancer pathogenesis. BIOIMPACTS : BI 2024; 15:30030. [PMID: 39963570 PMCID: PMC11830145 DOI: 10.34172/bi.30030] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/11/2023] [Accepted: 10/28/2023] [Indexed: 02/20/2025]
Abstract
Introduction Signal transducer and activator of transcriptions (STATs) factors as critical proteins in cell signaling regulate diverse biological processes such as differentiation and proliferation of cells. STATs have been shown to play distinct roles in modulating immune responses mediated by innate and adaptive immune cell subsets due to their significant roles in cytokine signaling. Methods In the current study, we review recent studies on the contribution of individual STAT proteins to cytokine signaling, development, and activity of diverse immune cells that constitute the whole immune system and help its performance against endogenous or exogenous agents with a particular focus on meaningful STAT factor in each of innate and adaptive immune cells' subsets to clarify their function in favor of the tumor or against it. Results Dysregulation of signaling pathways in the immune cells is associated with various immune disorders, such as the inability of immune system cells in the effective destruction of cancerous cells. Increase of knowledge about these pathways' functions is essential to understand how they can be effectively targeted to eliminate tumors. Conclusion The majority of immune cells use the Jak/STAT signaling pathway, which is one of the most important signaling pathways with a role in induction of proper immune responses. Since each of the STAT factors has a specific role in diverse immune cells' subsets, appropriate targeting of them can be a promising strategy for patients who suffer from immune system disorders; specifically it can be beneficial as an approach for cancer immunotherapy.
Collapse
Affiliation(s)
- Sepideh Sohrabi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shiva Alipour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Javad Masoumi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
2
|
Huang GX, Hallen NR, Lee M, Zheng K, Wang X, Mandanas MV, Djeddi S, Fernandez D, Hacker J, Ryan T, Bergmark RW, Bhattacharyya N, Lee S, Maxfield AZ, Roditi RE, Buchheit KM, Laidlaw TM, Gern JE, Hallstrand TS, Ray A, Wenzel SE, Boyce JA, Gutierrez-Arcelus M, Barrett NA. Increased epithelial mTORC1 activity in chronic rhinosinusitis with nasal polyps. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.13.562288. [PMID: 37904989 PMCID: PMC10614789 DOI: 10.1101/2023.10.13.562288] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Background The airway epithelium plays a central role in the pathogenesis of chronic respiratory diseases such as asthma and chronic rhinosinusitis with nasal polyps (CRSwNP), but the mechanisms by which airway epithelial cells (EpCs) maintain inflammation are poorly understood. Objective We hypothesized that transcriptomic assessment of sorted airway EpCs across the spectrum of differentiation would allow us to define mechanisms by which EpCs perpetuate airway inflammation. Methods Ethmoid sinus EpCs from adult patients with CRS were sorted into 3 subsets, bulk RNA sequenced, and analyzed for differentially expressed genes and pathways. Single cell RNA-seq (scRNA-seq) datasets from eosinophilic and non-eosinophilic CRSwNP and bulk RNA-seq of EpCs from mild/moderate and severe asthma were assessed. Immunofluorescent staining and ex vivo functional analysis of sinus EpCs were used to validate our findings. Results Analysis within and across purified EpC subsets revealed an enrichment in glycolytic programming in CRSwNP vs CRSsNP. Correlation analysis identified mammalian target of rapamycin complex 1 (mTORC1) as a potential regulator of the glycolytic program and identified EpC expression of cytokines and wound healing genes as potential sequelae. mTORC1 activity was upregulated in CRSwNP, and ex vivo inhibition demonstrated that mTOR is critical for EpC generation of CXCL8, IL-33, and CXCL2. Across patient samples, the degree of glycolytic activity was associated with T2 inflammation in CRSwNP, and with both T2 and non-T2 inflammation in severe asthma. Conclusions Together, these findings highlight a metabolic axis required to support epithelial generation of cytokines critical to both chronic T2 and non-T2 inflammation in CRSwNP and asthma.
Collapse
Affiliation(s)
- George X. Huang
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Nils R. Hallen
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Minkyu Lee
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Kelly Zheng
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Xin Wang
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | | | - Sarah Djeddi
- Division of Immunology, Boston Children’s Hospital; Boston, MA
| | | | - Jonathan Hacker
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Tessa Ryan
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Regan W. Bergmark
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women’s Hospital; Boston, MA
| | - Neil Bhattacharyya
- Department of Otolaryngology, Head and Neck Surgery, Massachusetts Eye and Ear Infirmary; Boston, MA
| | - Stella Lee
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women’s Hospital; Boston, MA
| | - Alice Z. Maxfield
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women’s Hospital; Boston, MA
| | - Rachel E. Roditi
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women’s Hospital; Boston, MA
| | - Kathleen M. Buchheit
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Tanya M. Laidlaw
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - James E. Gern
- Division of Allergy, Immunology, and Rheumatology, University of Wisconsin School of Medicine and Public Health; Madison, WI
| | - Teal S. Hallstrand
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington Medical Center; Seattle, WA
| | - Anuradha Ray
- Department of Immunology, University of Pittsburgh; Pittsburgh, PA
| | - Sally E. Wenzel
- Department of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center; Pittsburgh, PA
| | - Joshua A. Boyce
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Maria Gutierrez-Arcelus
- Division of Immunology, Boston Children’s Hospital; Boston, MA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard; Cambridge, MA
| | - Nora A. Barrett
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| |
Collapse
|
3
|
Gauthier M, Kale SL, Oriss TB, Gorry M, Ramonell RP, Dalton K, Ray P, Fahy JV, Seibold MA, Castro M, Jarjour N, Gaston B, Bleecker ER, Meyers DA, Moore W, Hastie AT, Israel E, Levy BD, Mauger D, Erzurum S, Comhair SA, Wenzel SE, Ray A. CCL5 is a potential bridge between type 1 and type 2 inflammation in asthma. J Allergy Clin Immunol 2023; 152:94-106.e12. [PMID: 36893862 PMCID: PMC10330021 DOI: 10.1016/j.jaci.2023.02.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 01/06/2023] [Accepted: 02/13/2023] [Indexed: 03/09/2023]
Abstract
BACKGROUND Type 1 (T1) inflammation (marked by IFN-γ expression) is now consistently identified in subsets of asthma cohorts, but how it contributes to disease remains unclear. OBJECTIVE We sought to understand the role of CCL5 in asthmatic T1 inflammation and how it interacts with both T1 and type 2 (T2) inflammation. METHODS CCL5, CXCL9, and CXCL10 messenger RNA expression from sputum bulk RNA sequencing, as well as clinical and inflammatory data were obtained from the Severe Asthma Research Program III (SARP III). CCL5 and IFNG expression from bronchoalveolar lavage cell bulk RNA sequencing was obtained from the Immune Mechanisms in Severe Asthma (IMSA) cohort and expression related to previously identified immune cell profiles. The role of CCL5 in tissue-resident memory T-cell (TRM) reactivation was evaluated in a T1high murine severe asthma model. RESULTS Sputum CCL5 expression strongly correlated with T1 chemokines (P < .001 for CXCL9 and CXCL10), consistent with a role in T1 inflammation. CCL5high participants had greater fractional exhaled nitric oxide (P = .009), blood eosinophils (P < .001), and sputum eosinophils (P = .001) in addition to sputum neutrophils (P = .001). Increased CCL5 bronchoalveolar lavage expression was unique to a previously described T1high/T2variable/lymphocytic patient group in the IMSA cohort, with IFNG trending with worsening lung obstruction only in this group (P = .083). In a murine model, high expression of the CCL5 receptor CCR5 was observed in TRMs and was consistent with a T1 signature. A role for CCL5 in TRM activation was supported by the ability of the CCR5 inhibitor maraviroc to blunt reactivation. CONCLUSION CCL5 appears to contribute to TRM-related T1 neutrophilic inflammation in asthma while paradoxically also correlating with T2 inflammation and with sputum eosinophilia.
Collapse
Affiliation(s)
- Marc Gauthier
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa.
| | - Sagar Laxman Kale
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Timothy B Oriss
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Michael Gorry
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Richard P Ramonell
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Kathryn Dalton
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Prabir Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - John V Fahy
- Division of Pulmonary Allergy and Critical Care, University of California, San Francisco, Calif
| | - Max A Seibold
- Center for Genes, Environment, and Health and Department of Pediatrics, National Jewish Health, Denver, Colo; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, Colo
| | - Mario Castro
- Pulmonary, Critical Care and Sleep Medicine, University of Kansas School of Medicine, Kansas City, Kan
| | - Nizar Jarjour
- Division of Allergy, Pulmonary and Critical Care Medicine, University of Wisconsin School of Medicine, Madison, Wis
| | - Benjamin Gaston
- Riley Hospital for Children and Indiana University School of Medicine Department of Pediatrics, Indianapolis, Ind
| | - Eugene R Bleecker
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, Ariz
| | - Deborah A Meyers
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, Ariz
| | - Wendy Moore
- Section on Pulmonary, Critical Care, Allergy & Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC
| | - Annette T Hastie
- Section on Pulmonary, Critical Care, Allergy & Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC
| | - Elliot Israel
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Bruce D Levy
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - David Mauger
- Division of Statistics and Bioinformatics, Department of Public Health Sciences, Pennsylvania State University, Hershey, Pa
| | - Serpil Erzurum
- Lerner Research Institute, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Suzy A Comhair
- Lerner Research Institute, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Sally E Wenzel
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa; Department of Environmental and Occupation Health, University of Pittsburgh School of Public Health, Pittsburgh, Pa
| | - Anuradha Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| |
Collapse
|
4
|
Zheng C, Shi Y, Zou Y. T cell co-stimulatory and co-inhibitory pathways in atopic dermatitis. Front Immunol 2023; 14:1081999. [PMID: 36993982 PMCID: PMC10040887 DOI: 10.3389/fimmu.2023.1081999] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/28/2023] [Indexed: 03/14/2023] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) targeting the T cell inhibitory pathways has revolutionized cancer treatment. However, ICIs might induce progressive atopic dermatitis (AD) by affecting T cell reactivation. The critical role of T cells in AD pathogenesis is widely known. T cell co-signaling pathways regulate T cell activation, where co-signaling molecules are essential for determining the magnitude of the T cell response to antigens. Given the increasing use of ICIs in cancer treatment, a timely overview of the role of T cell co-signaling molecules in AD is required. In this review, we emphasize the importance of these molecules involved in AD pathogenesis. We also discuss the potential of targeting T cell co-signaling pathways to treat AD and present the unresolved issues and existing limitations. A better understanding of the T cell co-signaling pathways would aid investigation of the mechanism, prognosis evaluation, and treatment of AD.
Collapse
Affiliation(s)
- Chunjiao Zheng
- Skin and Cosmetic Research Department, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuling Shi
- Institute of Psoriasis, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Yuling Shi, ; Ying Zou,
| | - Ying Zou
- Skin and Cosmetic Research Department, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Yuling Shi, ; Ying Zou,
| |
Collapse
|
5
|
Ray A, Das J, Wenzel SE. Determining asthma endotypes and outcomes: Complementing existing clinical practice with modern machine learning. Cell Rep Med 2022; 3:100857. [PMID: 36543110 PMCID: PMC9798025 DOI: 10.1016/j.xcrm.2022.100857] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 10/24/2022] [Accepted: 11/18/2022] [Indexed: 12/24/2022]
Abstract
There is unprecedented opportunity to use machine learning to integrate high-dimensional molecular data with clinical characteristics to accurately diagnose and manage disease. Asthma is a complex and heterogeneous disease and cannot be solely explained by an aberrant type 2 (T2) immune response. Available and emerging multi-omics datasets of asthma show dysregulation of different biological pathways including those linked to T2 mechanisms. While T2-directed biologics have been life changing for many patients, they have not proven effective for many others despite similar biomarker profiles. Thus, there is a great need to close this gap to understand asthma heterogeneity, which can be achieved by harnessing and integrating the rich multi-omics asthma datasets and the corresponding clinical data. This article presents a compendium of machine learning approaches that can be utilized to bridge the gap between predictive biomarkers and actual causal signatures that are validated in clinical trials to ultimately establish true asthma endotypes.
Collapse
Affiliation(s)
- Anuradha Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, 3459 Fifth Avenue, MUH 628 NW, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Jishnu Das
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Systems Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Sally E Wenzel
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, 3459 Fifth Avenue, MUH 628 NW, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Environmental Medicine and Occupational Health, School of Public Health, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
6
|
Tsiogka A, Kyriazopoulou M, Kontochristopoulos G, Nicolaidou E, Stratigos A, Rigopoulos D, Gregoriou S. The JAK/STAT Pathway and Its Selective Inhibition in the Treatment of Atopic Dermatitis: A Systematic Review. J Clin Med 2022; 11:jcm11154431. [PMID: 35956047 PMCID: PMC9369061 DOI: 10.3390/jcm11154431] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/19/2022] [Accepted: 07/27/2022] [Indexed: 02/04/2023] Open
Abstract
In recent years, the broadening understanding of the pathogenesis of atopic dermatitis (AD) has led to the development of novel therapeutic molecules, that target core inflammatory components of the disease. The Janus kinase (JAK)/signal transducer and activation of transcription (STAT) pathway constitutes the principal signaling cascade for a large number of cytokines and growth factors and is involved in intracellular signal transduction and subsequent regulation of gene transcription. Current knowledge suggests that the robust activation of the T-helper (Th)-2 [interleukin (IL)-4, IL-5, IL-13, IL-31] and Th22 (IL-22) immune responses in both skin and serum plays a pivotal role in the immunopathogenesis of AD especially at the acute stage, followed by a variable degree of Th1 (interferon-γ, tumor necrosis factor alpha) and Th17 (IL-17) activation in chronic disease. Of note, most of the aforementioned inflammatory cytokines utilize the JAK/STAT pathway for downstream signal transduction, explaining the emerging role of JAK inhibitors in the therapeutic armamentarium of AD. The present systematic review aims to discuss the involvement of JAK/STAT pathway in the pathogenesis of AD and summarize the clinical data available on the efficacy and safety of JAK inhibitors which have been used in the treatment of AD thus far.
Collapse
|
7
|
Sailliet N, Brosseau C, Robert JM, Brouard S. Role of JAK inhibitors and immune cells in transplantation. Cytokine Growth Factor Rev 2019; 47:62-73. [DOI: 10.1016/j.cytogfr.2019.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 05/09/2019] [Indexed: 02/07/2023]
|
8
|
Hagmann BR, Odermatt A, Kaufmann T, Dahinden CA, Fux M. Balance between IL-3 and type Iinterferons and their interrelationship with FasL dictates lifespan and effector functions of human basophils. Clin Exp Allergy 2016; 47:71-84. [PMID: 27910206 DOI: 10.1111/cea.12850] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 09/13/2016] [Accepted: 10/10/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND In contrast to eosinophils and neutrophils, the regulation of the lifespan of human basophils is poorly defined, with the exception of the potent anti-apoptotic effect of IL-3 that also promotes pro-inflammatory effector functions and phenotypic changes. Type I IFNs (IFN-α, IFN-β), which are well known for their anti-viral activities, have the capacity to inhibit allergic inflammation. OBJECTIVE To elucidate whether type I IFNs have the potential to abrogate the lifespan and/or effector functions of human basophils. METHODS We cultured human basophils, and for comparison, eosinophils and neutrophils, with IL-3, interferons, FasL and TRAIL, alone or in combination, and studied cell survival, effector functions and signalling pathways involved. RESULTS Despite an identical pattern of early signalling in basophils, eosinophils and neutrophils in response to different types of interferons, only basophils displayed enhanced apoptosis after type I IFN treatment. IFN-γ prolonged survival of eosinophils but did not affect the lifespan of basophils. IFN-α-mediated apoptosis required STAT1-STAT2 heterodimers and the contribution of constitutive p38 MAPK activity. Whereas the death ligands FasL and TRAIL-induced apoptosis in basophils per se, IFN-α-mediated apoptosis did neither involve autocrine TRAIL signalling nor did it sensitize basophils to FasL-induced apoptosis. However, IFN-α and FasL displayed an additive effect in killing basophils. Interestingly, IL-3, which protected basophils from IFN-α-, TRAIL- or FasL-mediated apoptosis, did not completely block the additive effect of combined IFN-α and FasL treatment. Moreover, we demonstrate that IFN-α suppressed IL-3-induced release of IL-8 and IL-13. In contrast to IFN-α-mediated apoptosis, these inhibitory effects of IFN-α were not dependent on p38 MAPK signalling. CONCLUSIONS AND CLINICAL RELEVANCE Our study defines the unique and granulocyte-type-specific inhibitory and pro-apoptotic function of type I IFNs and their cooperation with death ligands in human blood basophils, which may be relevant for the anti-allergic properties of type I IFNs.
Collapse
Affiliation(s)
- B R Hagmann
- University Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,University Institute of Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - A Odermatt
- University Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,University Institute of Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - T Kaufmann
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - C A Dahinden
- University Institute of Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - M Fux
- University Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,University Institute of Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
9
|
Shen ZJ, Malter JS. Determinants of eosinophil survival and apoptotic cell death. Apoptosis 2015; 20:224-34. [PMID: 25563855 DOI: 10.1007/s10495-014-1072-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Eosinophils (Eos) are potent inflammatory cells and abundantly present in the sputum and lung of patients with allergic asthma. During both transit to and residence in the lung, Eos contact prosurvival cytokines, particularly IL-3, IL-5 and GM-CSF, that attenuate cell death. Cytokine signaling modulates the expression and function of a number of intracellular pro- and anti-apoptotic molecules. Both intrinsic mitochondrial and extrinsic receptor-mediated pathways are affected. This article discusses the fundamental role of the extracellular and intracellular molecules that initiate and control survival decisions by human Eos and highlights the role of the cis-trans isomerase, Pin1 in controlling these processes.
Collapse
Affiliation(s)
- Zhong-Jian Shen
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-9072, USA,
| | | |
Collapse
|
10
|
Levy LL, Urban J, King BA. Treatment of recalcitrant atopic dermatitis with the oral Janus kinase inhibitor tofacitinib citrate. J Am Acad Dermatol 2015; 73:395-9. [PMID: 26194706 DOI: 10.1016/j.jaad.2015.06.045] [Citation(s) in RCA: 153] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 06/16/2015] [Accepted: 06/24/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND Treatment of moderate to severe atopic dermatitis (AD) is often inadequate. OBJECTIVE We sought to evaluate the efficacy of the oral Janus kinase inhibitor tofacitinib citrate in the treatment of moderate to severe AD. METHODS Six consecutive patients with moderate to severe AD who had failed standard treatment were treated with tofacitinib citrate. Response to treatment was assessed using the Scoring of AD index. RESULTS Decreased body surface area involvement of dermatitis and decreased erythema, edema/papulation, lichenification, and excoriation were observed in all patients. The Scoring of AD index decreased by 66.6% from 36.5 to 12.2 (P < .05) during 8 to 29 weeks of treatment. There were no adverse events. LIMITATIONS Small sample size, lack of placebo control group, and the possibility of bias are limitations. CONCLUSION The oral Janus kinase inhibitor tofacitinib citrate may be beneficial in the treatment of moderate to severe AD.
Collapse
Affiliation(s)
- Lauren L Levy
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut
| | - Jennifer Urban
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut
| | - Brett A King
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
11
|
Bao L, Zhang H, Chan LS. The involvement of the JAK-STAT signaling pathway in chronic inflammatory skin disease atopic dermatitis. JAKSTAT 2013; 2:e24137. [PMID: 24069552 PMCID: PMC3772104 DOI: 10.4161/jkst.24137] [Citation(s) in RCA: 163] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Revised: 02/26/2013] [Accepted: 02/27/2013] [Indexed: 12/18/2022] Open
Abstract
Atopic dermatitis (AD), a common chronic inflammatory skin disease, is characterized by inflammatory cell skin infiltration. The JAK-STAT pathway has been shown to play an essential role in the dysregulation of immune responses in AD, including the exaggeration of Th2 cell response, the activation of eosinophils, the maturation of B cells, and the suppression of regulatory T cells (Tregs). In addition, the JAK-STAT pathway, activated by IL-4, also plays a critical role in the pathogenesis of AD by upregulating epidermal chemokines, pro-inflammatroy cytokines, and pro-angiogenic factors as well as by downregulating antimicrobial peptides (AMPs) and factors responsible for skin barrier function. In this review, we will highlight the recent advances in our understanding of the JAK-STAT pathway in the pathogenesis of AD.
Collapse
Affiliation(s)
- Lei Bao
- Department of Dermatology; University of Illinois; Chicago, IL USA
| | | | | |
Collapse
|
12
|
Abstract
Accurate diagnosis of eosinophilic lung diseases is essential to optimizing patient outcomes, but remains challenging. Signs and symptoms frequently overlap among the disorders, and because these disorders are infrequent, expertise is difficult to acquire. Still, these disorders are not rare, and most clinicians periodically encounter patients with one or more of the eosinophilic lung diseases and need to understand how to recognize, diagnose, and manage these diseases. This review focuses on the clinical features, general diagnostic workup, and management of the eosinophilic lung diseases.
Collapse
Affiliation(s)
- Evans R Fernández Pérez
- Interstitial Lung Disease Program, Autoimmune Lung Center, National Jewish Health, Denver, CO 80206, USA.
| | | | | |
Collapse
|
13
|
Kanda A, Driss V, Hornez N, Abdallah M, Roumier T, Abboud G, Legrand F, Staumont-Sallé D, Quéant S, Bertout J, Fleury S, Rémy P, Papin JP, Julia V, Capron M, Dombrowicz D. Eosinophil-derived IFN-gamma induces airway hyperresponsiveness and lung inflammation in the absence of lymphocytes. J Allergy Clin Immunol 2009; 124:573-82, 582.e1-9. [PMID: 19539982 DOI: 10.1016/j.jaci.2009.04.031] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2008] [Revised: 04/22/2009] [Accepted: 04/24/2009] [Indexed: 01/21/2023]
Abstract
BACKGROUND Eosinophils are key players in T(H)2-driven pathologies, such as allergic lung inflammation. After IL-5- and eotaxin-mediated tissue recruitment, they release several cytotoxic and inflammatory mediators. However, their exact contribution to asthma remains controversial. Indeed, in human subjects anti-IL-5 treatment inhibits eosinophilia but not antigen-induced airway hyperresponsiveness (AHR). Likewise, lung fibrosis is abrogated in 2 strains of eosinophil-deficient mice, whereas AHR is inhibited in only one of them. Finally, eosinophils have been shown to attract T(H)2 lymphocytes at the inflammatory site. OBJECTIVE The ability of eosinophils to promote AHR and lung inflammation independently of lymphocytes was investigated. METHODS Adoptive transfers of resting or activated eosinophils from IL-5 transgenic mice were performed into naive BALB/c mice, mice with severe combined immunodeficiency, and IFN-gamma-deficient BALB/c recipients. RESULTS Adoptively transferred eosinophils induced lung inflammation, fibrosis, collagen deposition, and AHR not only in BALB/c mice but also in recipient mice with severe combined immunodeficiency. Surprisingly, IFN-gamma expression was increased in lungs from eosinophil-transferred animals. Furthermore, IFN-gamma neutralization in recipients partially inhibited eosinophil-induced AHR. Moreover, IFN-gamma-deficient eosinophils or eosinophils treated with a blocking anti-IFN-gamma receptor antibody failed to induce AHR in IFN-gamma-deficient recipients. Finally, in vitro and at low concentrations, IFN-gamma increased eosinophil peroxidase release, potentiated chemotaxis, and prolonged survival, suggesting the existence of an autocrine mechanism. CONCLUSIONS These results support the important and previously unsuspected contribution of eosinophils to lung inflammation independently of lymphocytes through production of IFN-gamma, the prototypical T(H)1 cytokine.
Collapse
Affiliation(s)
- Akira Kanda
- Inserm U547, Lille, France; Institut Pasteur de Lille, Lille, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Matsuwaki Y, Ueki S, Adachi T, Oyamada H, Kamada Y, Yamaguchi K, Kanda A, Hamada K, Kayaba H, Chihara J. The synthetic PPARgamma agonist troglitazone inhibits IL-5-induced CD69 upregulation and eosinophil-derived neurotoxin release from eosinophils. Pharmacology 2005; 74:169-73. [PMID: 15818060 DOI: 10.1159/000085034] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2004] [Accepted: 02/02/2005] [Indexed: 11/19/2022]
Abstract
Peroxisome proliferator-activated receptor-gamma (PPARgamma) is a nuclear receptor that regulates lipid metabolism. Recently, PPARgamma was reported to be a negative regulator in the immune system. Eosinophils also express PPARgamma, however, the role of PPARgamma in eosinophil functions is not well understood. Surface expression of CD69 and eosinophil-derived neurotoxin (EDN) release are well-known activation markers of eosinophils. We investigated the effect of a PPARgamma agonist on human eosinophil functions such as IL-5-induced CD69 surface expression and EDN release. IL-5 significantly induced eosinophil CD69 surface expression analyzed using flow cytometry and EDN release measured by ELISA. IL-5-induced eosinophil CD69 surface expression and EDN release were significantly inhibited by the synthetic PPARgamma agonist troglitazone, and these effects were reversed by a PPARgamma antagonist. The PPARgamma agonist troglitazone has a potent inhibitory effect on activation and degranulation of eosinophils, and it may be a therapeutic modality for the treatment of allergic diseases.
Collapse
Affiliation(s)
- Yoshinori Matsuwaki
- Department of Clinical and Laboratory Medicine, Akita University School of Medicine, Akita, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Kondo Y, Matsuse H, Machida I, Kawano T, Saeki S, Tomari S, Obase Y, Fukushima C, Kohno S. Effects of primary and secondary low-grade respiratory syncytial virus infections in a murine model of asthma. Clin Exp Allergy 2004; 34:1307-13. [PMID: 15298574 DOI: 10.1111/j.1365-2222.2004.02033.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infection is known to develop and exacerbate asthma in young children. In adult, RSV causes recurrent but asymptomatic infections. However, the impact of asymptomatic RSV infection on adult asthma is yet to be determined. The present study is designed to determine the effects of primary and secondary low-grade RSV infections on allergic airway inflammation in a murine model of allergic asthma. METHODS A low-grade RSV (2 x 10(3) plaque-forming units/mouse) was inoculated, and this caused neither pulmonary inflammation nor symptoms but induced significant IFN-gamma production in thoracic lymph nodes. To investigate interaction between low-grade virus and Dermatophagoides farinae (Df), airway hyper-responsiveness, lung inflammation and cytokine production from thoracic lymph nodes were compared after primary and secondary low-grade RSV infections in four groups of mice; control, Df allergen-sensitized, RSV-infected and Df-sensitized RSV-infected mice. A direct comparison between low- and high-grade RSV infections was also performed in primary infection. To investigate the role of IL-5 during secondary RSV infection, anti-IL-5 monoclonal antibody (anti-IL-5 mAb) was injected in mice and similar parameters were compared in four groups of mice. RESULTS Primary high-grade RSV infection increased allergen-induced airway inflammation, while primary low-grade RSV infection attenuated allergen-induced airway inflammation concomitant with significant IFN-gamma production in lung-draining lymph nodes. In marked contrast, secondary low-grade RSV infection increased both IFN-gamma and IL-5 production, resulting in exacerbation of allergen-induced airway inflammation. Anti-IL-5 mAb treatment in secondary low-grade RSV infection and Df allergen-sensitized mice attenuated virus and allergen-induced airway inflammation. CONCLUSIONS Low-grade RSV infection per se does not cause pulmonary inflammation, whereas it induces a significant immunological response in the allergen-sensitized host. These results indicate that subclinical and recurrent RSV infection may play an important role in exacerbation and maintenance of asthma in adults, wherein IL-5 is critically involved.
Collapse
Affiliation(s)
- Y Kondo
- Second Department of Internal Medicine, Nagasaki University School of Medicine, Nagasaki, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Rendon-Mitchell B, Ochani M, Li J, Han J, Wang H, Yang H, Susarla S, Czura C, Mitchell RA, Chen G, Sama AE, Tracey KJ, Wang H. IFN-gamma induces high mobility group box 1 protein release partly through a TNF-dependent mechanism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:3890-7. [PMID: 12646658 DOI: 10.4049/jimmunol.170.7.3890] [Citation(s) in RCA: 269] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We recently discovered that a ubiquitous protein, high mobility group box 1 protein (HMGB1), is released by activated macrophages, and functions as a late mediator of lethal systemic inflammation. To elucidate mechanisms underlying the regulation of HMGB1 release, we examined the roles of other cytokines in induction of HMGB1 release in macrophage cell cultures. Macrophage migration inhibitory factor, macrophage-inflammatory protein 1beta, and IL-6 each failed to significantly induce the release of HMGB1 even at supraphysiological levels (up to 200 ng/ml). IFN-gamma, an immunoregulatory cytokine known to mediate the innate immune response, dose-dependently induced the release of HMGB1, TNF, and NO, but not other cytokines such as IL-1alpha, IL-1beta, or IL-6. Pharmacological suppression of TNF activity with neutralizing Abs, or genetic disruption of TNF expression (TNF knockout) partially (50-60%) inhibited IFN-gamma-mediated HMGB1 release. AG490, a specific inhibitor for Janus kinase 2 of the IFN-gamma signaling pathway, dose-dependently attenuated IFN-gamma-induced HMGB1 release. These data suggest that IFN-gamma plays an important role in the regulation of HMGB1 release through a TNF- and Janus kinase 2-dependent mechanism.
Collapse
Affiliation(s)
- Beatriz Rendon-Mitchell
- Center of Immunology and Inflammation, North Shore-Long Island Jewish Research Institute, Manhasset, NY 11030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Becler K, Håkansson L, Rak S. Treatment of asthmatic patients with a cysteinyl leukotriene receptor-1 antagonist montelukast (Singulair), decreases the eosinophil survival-enhancing activity produced by peripheral blood mononuclear leukocytes in vitro. Allergy 2002; 57:1021-8. [PMID: 12358998 DOI: 10.1034/j.1398-9995.2002.23620.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Montelukast (Singulair, MSD) has been shown to have a beneficial effect on the clinical symptoms of asthma. We aimed to investigate the effect of montelukast treatment on the production of eosinophil survival-enhancing cytokines by peripheral blood mononuclear cells (PBMNC). METHODS PBMNC obtained from 15 grass-allergic patients (7 treated with montelukast and 8 with a placebo) were cultured for 72 h. Eosinophils from allergic patients were cultured with MNC supernatants alone or with addition of neutralizing antibodies, and the proportion of living cells was assessed by flow cytometry. In another experiment PBMNC from 6 allergic patients were cultured in vitro in the presence of montelukast or vehicle. Following stimulation the production of GM-CSF in monocytes was assessed. RESULTS Eosinophil survival in the MNC supernatants from the placebo-treated patients was significantly (P < 0.05) higher than in supernatants from montelukast-treated patients. GM-CSF was the predominant cytokine responsible for the eosinophil survival-enhancing activity (ESEA). In vitro production of GM-CSF by allergen-stimulated monocytes was significantly suppressed by addition of montelukast. CONCLUSION Treatment of patients with montelukast decreased the production of MNC-derived cytokines, particularly GM-CSF. We suggest that cysteinyl leukotriene receptor-1 (CysLT-R1) antagonists may act, at least partially, by diminishing the production of GM-CSF from PBMNCs.
Collapse
Affiliation(s)
- K Becler
- Asthma and Allergy Research Group, Department of Respiratory Medicine and Allergology, Sahlgrenska University Hospital, Bruna stråket 11, 413 45 Göteborg, Sweden
| | | | | |
Collapse
|
18
|
Zheng X, Karsan A, Duronio V, Chu F, Walker DC, Bai TR, Schellenberg RR. Interleukin-3, but not granulocyte-macrophage colony-stimulating factor and interleukin-5, inhibits apoptosis of human basophils through phosphatidylinositol 3-kinase: requirement of NF-kappaB-dependent and -independent pathways. Immunology 2002; 107:306-15. [PMID: 12423306 PMCID: PMC1782810 DOI: 10.1046/j.1365-2567.2002.01517.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Basophils are key effector cells of allergic reactions. Although proinflammatory cytokines, such as interleukin (IL)-3, granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL-5, inhibit eosinophil apoptosis in vitro, little is known about basophil apoptosis, and the signalling mechanisms required for basophil survival remain undefined. To address this issue, we used a novel negative-selection system to isolate human basophils to a purity of > 95%, and evaluated apoptosis by morphology using light and transmission electron microscopy, and by annexin-V binding and propidium iodide incorporation using flow cytometry. In this study, we demonstrated that the spontaneous rate of apoptotic basophils was higher than that of eosinophils as, at 24 hr, 57.6 +/- 4.7% of basophils underwent apoptosis compared with 39.5 +/- 3.8% of eosinophils. In addition, basophil cell death was significantly inhibited when cultured with IL-3 for 48 hr (84.6 +/- 4.9% vehicle-treated cells versus 40.9 +/- 3.9% IL-3-treated cells). IL-3 also up-regulated basophil CD69 surface expression. The effects of IL-3 on apoptosis and CD69 surface expression of human basophils were completely blocked by LY294002 (LY), a potent inhibitor of phosphatidylinositol 3-kinase (PI3-K), but only partially inhibited by lactacystin, a proteasome inhibitor that prevents degradation of IkappaB and NF-kappaB translocation. These observations reveal the novel finding that IL-3 prevents basophil apoptosis through the activation of PI3-K, which is only partially NF-kappaB dependent. As basophils are active participants in allergic reactions and IL-3 is one of the abundant proinflammatory cytokines in secretions from allergic tissue, we suggest that IL-3-mediated inhibition of basophil apoptosis may exacerbate the inflammation associated with allergic disorders.
Collapse
Affiliation(s)
- Xueyan Zheng
- iCAPTURE Center, McDonald Research Laboratory, University of British Columbia, St. Paul's Hospital, 1081 Burrard Street, Vancouver, BC, Canada V6Z 1Y6
| | | | | | | | | | | | | |
Collapse
|
19
|
Riffo-Vasquez Y, Spina D. Role of cytokines and chemokines in bronchial hyperresponsiveness and airway inflammation. Pharmacol Ther 2002; 94:185-211. [PMID: 12113798 DOI: 10.1016/s0163-7258(02)00217-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Over the last decade there has been an intense interest in the potential role of cytokines and chemokines as important mediators in various atopic diseases, including asthma and the mechanisms by which these mediators regulate airway inflammation and bronchial hyperresponsiveness. This research effort has recently culminated in the publication of clinical studies that have assessed the role of interleukin (IL)-4 [Borish et al., Am J Respir Crit Care Med 160, 1816-1823 (1999)], IL-5 [Leckie et al., Lancet 356, 2144-2148 (2000)], and IL-12 [Bryan et al., Lancet 356, 2149-2153 (2000)] in allergic asthma, and the results have been disappointing. This is not surprising given the pleiotropic role cytokines play in the allergic response confirmed by numerous animal studies providing evidence of functional redundancy. The alternative view is that our current concepts in asthma pathogenesis need significant revision. This review will summarise the evidence for the role of cytokines and chemokines in various aspects of asthma pathophysiology; namely, bronchial hyperresponsiveness, eosinophil recruitment to the airways, mucus secretion, and airway remodelling.
Collapse
Affiliation(s)
- Y Riffo-Vasquez
- The Sacker Institute of Pulmonary Pharmacology, 5th Floor Hodgkin Building, GKT School of Biomedical Science, King's College London, London SE1 1UL, UK
| | | |
Collapse
|
20
|
Mohanty JG, Raible DG, McDermott LJ, Pelleg A, Schulman ES. Effects of purine and pyrimidine nucleotides on intracellular Ca2+ in human eosinophils: activation of purinergic P2Y receptors. J Allergy Clin Immunol 2001; 107:849-55. [PMID: 11344352 DOI: 10.1067/mai.2001.114658] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND Extracellular adenosine 5'-triphosphate (ATP) increases human eosinophil intracellular Ca(2+) concentration; the mechanism of action is not fully known. ATP, a physiologic regulator, acts through 2 purinergic receptor types: cation channels (P2X) and G protein-coupled receptors (P2Y). OBJECTIVE This study is aimed at identifying the functional purinergic receptors in human eosinophils. METHODS The relative potency of ATP, uridine (UTP), cytidine (CTP), and inosine (ITP) 5'-triphosphates (P2Y agonists); 2-methylthio-ATP (P2Y(1) agonist); and 2 P2X agonists, alpha,beta-methylene-ATP and beta,gamma-methylene-ATP on intracellular Ca(2+) concentration was examined in Ca(2+)-sensitive Fura-2-labeled human eosinophils. For comparison, ATP effects were similarly studied in human neutrophils. P2X/P2Y mRNA expression in cells was examined by reverse transcription and PCR. RESULTS The nucleotide potency order was UTP > or = ATP > ITP >>> 2-methylthio-ATP > alpha,beta-methylene-ATP = beta,gamma-methylene-ATP = CTP = 0 in eosinophils. Pertussis toxin (500 ng/mL) pretreatment abolished the effect of lower (10(-6) mol/L) but not higher (10(-5) mol/L) concentrations of ATP in eosinophils, whereas it attenuated the effects of 10(-4) mol/L ATP in neutrophils. The phospholipase C inhibitor U73122 (2 micromol/L) partially inhibited the effect of ATP in eosinophils but totally blocked it in neutrophils. Both cells constitutively express mRNA for P2X(1), P2X(4), P2X(5), P2Y(1), and P2Y(2), but not P2X(7), with much weaker expressions of P2X(4) and P2X(5) in neutrophils. Eosinophils cultured with the T(H)1 cytokine, IFN-gamma, expressed mRNA for P2X(7), a receptor linked to apoptosis. CONCLUSIONS These results suggest that the P2 purinergic receptor signal transduction pathways in eosinophils and neutrophils are different and are mediated by more than 1 subtype of functional P2Y receptors.
Collapse
Affiliation(s)
- J G Mohanty
- Division of Pulmonary/Critical Care, Department of Medicine, MCP Hahnemann University, Philadelphia 19102, USA
| | | | | | | | | |
Collapse
|
21
|
Abstract
With over 50 potential asthma mediators, cytokines are the latest group of substances which have been investigated for their potential role in this disease. The use of murine models of allergic inflammation has facilitated the investigation of the role of individual cytokines in this response. The use of targeted gene disruption, overexpression of genes and monoclonal antibodies directed against cytokines have allowed a detailed examination of the role cytokines play in IgE production, eosinophil recruitment and bronchial hyperresponsiveness, which are the characteristic features of the asthma phenotype. Despite the introduction of this new methodology, conflicting reports relating to the role of cytokines in allergic inflammation, highlight the complexity of allergic inflammation and challenge the notion that a single cytokine can explain the asthma phenotype.
Collapse
Affiliation(s)
- Y Riffo-Vasquez
- The Sackler Institute of Pulmonary Pharmacology, GKT School of Biomedical Science, King's College, London, UK
| | | | | |
Collapse
|
22
|
Matsuse H, Behera AK, Kumar M, Lockey RF, Mohapatra SS. Differential cytokine mRNA expression in Dermatophagoides farinae allergen-sensitized and respiratory syncytial virus-infected mice. Microbes Infect 2000; 2:753-9. [PMID: 10955955 DOI: 10.1016/s1286-4579(00)00500-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The interaction between mite allergen sensitization and respiratory syncytial virus (RSV) infection at the level of cytokine mRNA expression was examined in a murine model in the present study. Primary RSV infection enhances expression of inflammatory cytokines such as IL-6, IFN-gamma, and eotaxin in the lung and upregulates the expression of Th2-like cytokines IL-10 and IL-13 in the spleen in BALB/c mice. Mite antigen-sensitized and RSV-infected (ASRSV) mice show enhanced (P < 0.05) total serum IgE compared to antigen-sensitized mice. However, the levels of viral mRNA in the lung tissues are comparable between RSV-infected and ASRSV mice. It is concluded that compartmentalization of cytokine expression following RSV infection plays a role in the augmentation of Th2-like and IgE antibody response to RSV.
Collapse
Affiliation(s)
- H Matsuse
- The Joy McCann Culverhouse Airway Disease Center, Department of Internal Medicine, University of South Florida and James A. Haley VA Hospital, Tampa 33612, USA
| | | | | | | | | |
Collapse
|
23
|
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease that frequently predates the development of allergic rhinitis or asthma. It is an important skin condition with significant costs and morbidity to patients and their families; the disease affects more than 10% of children. Recent studies have demonstrated the complex interrelationship of genetic, environmental, skin barrier, pharmacologic, psychologic, and immunologic factors that contribute to the development and severity of AD. The current review will examine the cellular and molecular mechanisms that contribute to AD as well as the immunologic triggers involved in its pathogenesis. These insights provide new opportunities for therapeutic intervention in this common skin condition.
Collapse
Affiliation(s)
- D Y Leung
- Division of Pediatric Allergy-Immunology, National Jewish Medical and Research Center, and the Department of Pediatrics, University of Colorado Health Sciences Center, Denver, CO 80206, USA
| |
Collapse
|